EP1346033A2 - Nouveaux genes de serine protease apparentes a la dppiv - Google Patents

Nouveaux genes de serine protease apparentes a la dppiv

Info

Publication number
EP1346033A2
EP1346033A2 EP01981501A EP01981501A EP1346033A2 EP 1346033 A2 EP1346033 A2 EP 1346033A2 EP 01981501 A EP01981501 A EP 01981501A EP 01981501 A EP01981501 A EP 01981501A EP 1346033 A2 EP1346033 A2 EP 1346033A2
Authority
EP
European Patent Office
Prior art keywords
dprp
seq
polypeptide
mature protein
nos
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
EP01981501A
Other languages
German (de)
English (en)
Inventor
Steve Qi
Karen O. Akinsanya
Pierre J.-M. Riviere
Jean-Louis Junien
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ferring BV
Original Assignee
Ferring BV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ferring BV filed Critical Ferring BV
Publication of EP1346033A2 publication Critical patent/EP1346033A2/fr
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/52Genes encoding for enzymes or proenzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/026Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a baculovirus

Definitions

  • the present invention relates to novel serine proteases related to dipeptidyl peptidase IV (DPPIN), and to isolated nucleic acids coding for these proteases, all of which are useful for the discovery of new therapeutic agents, for measuring protease activity, and for determining the inhibitory activity of compounds against these proteases.
  • DPPIN dipeptidyl peptidase IV
  • Proteases and peptidases are enzymes that catalyse the hydrolysis of peptidic amide bonds.
  • Proteases play an important role in the regulation of biological processes in almost every life-form from bacteria to virus to mammals. They perform critical functions in, for example, digestion, blood clotting, apoptosis, activation of immune responses, zymogen activation, viral maturation, protein secretion and protein trafficking. They can be classified according to a number of criteria, such as site of action, substrate preference, and mechanism.
  • aminopeptidases act preferentially at the ⁇ -terminal residues of a peptide
  • carboxypeptidases act preferentially at the C-terminus
  • endopeptidases act at sites removed from the two termini.
  • carboxy- and aminopeptidases cleave a single amino acid residue from the substrate
  • dipeptidyl peptidases cleave a dipeptide unit (two amino acids) from the substrate
  • tripeptidases cleave three amino acids from the substrate. Substrate preference is frequently expressed in terms of the amino acid residue immediately ⁇ -terminal to the cleavage site.
  • trypsin-like peptidases will preferentially cleave a peptide next to a basic amino acid (arginine or lysine), i.e. where the bond hydrolysed is the Arg/Lys-Xaa bond.
  • a basic amino acid arginine or lysine
  • the chymotrypsin-like family of peptidases preferentially hydrolyse peptides adjacent to an aromatic residue.
  • peptidases are classified as being serine- dependent, cysteine-dependent, aspartic acid-dependent or zinc-dependent.
  • proteases and proteases are involved in the regulation of many physiological processes, they are attractive targets for the development of therapeutic agents.
  • Protease and peptidase inhibitors are, for example, used in the treatment of hypertension, coagulation disorders, and viral infection.
  • Proteolytic enzymes that exploit serine in their catalytic activity are ubiquitous, being found in viruses, bacteria and eukaryotes.
  • Over 20 families (denoted SI - S27) of serine protease have been identified; these are grouped into 6 clans (SA, SB, SC, SE, SF and SG) on the basis of structural similarity and other functional evidence. Structures are known for four of the clans (SA, SB, SC and SE); these appear to be totally unrelated, suggesting at least four evolutionary origins of serine peptidases and possibly many more, Rawlings and Barrett, Meth. Enzvmol. 244: 19-61 (1994).
  • the prolyl oligopeptidase family consists of a number of evolutionarily related peptidases whose catalytic activity seems to be provided by a charge relay system similar to that of the trypsin family of serine proteases, but which evolved by independent convergent evolution.
  • a conserved serine residue has been shown experimentally (in E. coli protease II as well as in pig and bacterial PE) to be necessary for the catalytic mechanism.
  • This serine which is part of the catalytic triad (Ser, His, Asp), is generally located about 150 residues away from the C-terminal extremity of these enzymes (which are all proteins that contains about 700 to 800 amino acids).
  • dipeptidyl peptidase IN DPPIN, EC 3.414.5
  • type II glycoprotein DPPIN, EC 3.414.5
  • dipeptidyl aminopeptidases are characterised by their ability to cleave ⁇ -terminal dipeptides from a variety of small peptides. Dipeptidyl aminopeptidases show different substrate specificities and cellular localisation, suggesting different functions of each activity in peptide processing.
  • DPPIN is characterised by its capacity to cleave ⁇ - terminal dipeptides containing proline or alanine as the penultimate residue.
  • the DPPIN gene spans approximately 70 kb and contains 26 exons, ranging in size from 45 bp to 1.4 kb.
  • the nucleotide sequence (3,465 bp) of the cD ⁇ A contains an open reading frame encoding a polypeptide comprising 766 amino acids.
  • the nucleotides that encode the active site sequence are split between 2 exons. This clearly distinguishes the genomic organisation of the prolyl oligopeptidase family from that of the classic serine protease family.
  • DPPIV is widely distributed in mammalian tissues and is found in great abundance in the kidney, intestinal epithelium and placenta (Yaron, A. and ⁇ aider, F., Critical Reviews in Biochem. Mol. Biol. 1993 [1], 31).
  • the enzyme is expressed almost exclusively by activated T-lymphocytes of the CD4 + type where the enzyme has been shown to be synonymous with the cell-surface antigen CD26.
  • the exact role of DP-IN in human physiology is still not completely understood, recent research has shown that the enzyme clearly has a major role in human physiology and pathophysiology.
  • DPPIV expression appears late in thymic differentiation and is preferentially restricted to the CD4 + helper/memory population, and CD26 can deliver a potent co-stimulatory T-cell activation signal.
  • DPPIV also known as T-cell activation antigen CD26, therefore plays an important role in the immune response via association with CD45 tyrosine phosphatase and, through its ability to bind adenosine deaminase (ADA) to the T-cell surface, protects the T-cell from adenosine-mediated inhibition of proliferation.
  • ADA adenosine deaminase
  • the regulation of the function of chemokines by CD26/DPPIV appears to be essential for lymphocyte trafficking and infectivity of HIV strains.
  • DPPIV has been associated with numerous functions including involvement in T-cell activation, cell adhesion, digestion of proline containing peptides in the kidney and intestines, HIV infection and apoptosis, and regulation of tumorigenicity in certain melanoma cells, Pethiyagoda et al., Clin. Exp. Metastasis 2000:18(5 ⁇ :391-400.
  • DPPIV is also implicated in the endocrine regulation and metabolic physiology. More particularly, DPPIV cleaves the amino-terminal His- Ala dipeptide of GLP-1, generating a GLP-1 receptor antagonist, and thereby shortens the physiological response to GLP-1.
  • GLP-1 Glucagon-like peptide- 1
  • DPPIV Glucagon-like peptide- 1
  • Inhibitors of DPPIV are currently being studied in the clinic as potential therapeutic agents for type 2 diabetes and impaired glucose tolerance.
  • DPPIV In addition to, but independent of, its serine type catalytic activity, DPPIV binds closely to the soluble extracellular enzyme adenosine deaminase (ADA), acting as a receptor and is thought to mediate signal transduction.
  • ADA adenosine deaminase
  • DPPIV structure is characterized by two extracellular domains, an ⁇ / ⁇ fold hydrolase domain and a 7-blade beta-propeller domain consisting of repeated beta sheets of about 50 amino acids. Recently it has been shown that, besides selecting substrates by size, the beta-propeller domain, containing 10 of the 12 highly conserved cysteine residues, contributes to catalysis of the peptidase domain. In addition, the cysteine-rich domain is responsible for DPPIV-binding to collagen I and to extracellular ADA.
  • ADA adenosine deaminase
  • DPPIV is also reported to play a role in fibronectin- mediated interactions of cells with extracellular matrix. Recent studies show that the protease activity of DPPIV is not required for its anti-invasive activity because mutants of DPPIV that lack the extracellular serine protease activity maintain such activity.
  • DPPIV-I DPP-II
  • DPP-III DPP-III
  • DPP-X fibroblast activation protein
  • FAP fibroblast activation protein
  • the present invention provides proteins with prolyloligopeptidase (post-proline cleaving) activities that constitute three novel members of a family of proteins related to DPPIV, including the full-length proteins, alternative splice forms, subunits, and mutants, as well as nucleotide sequences encoding the same.
  • the present invention also provides methods of screening for substrates, interacting proteins, agonists, antagonists or inhibitors of the above proteins, and furthermore to pharmaceutical compositions comprising the proteins and/or mutants, derivatives and/or analogues thereof and/or ligands thereto.
  • dipeptidyl peptidase IV-related protein- 1, 2 & 3 DPRP-1, DPRP-2 and DPRP-3.
  • the amino acid sequences of DPRP-1, DPRP-2 and DPRP-3 are given in SEQ. ID NOS:l, 3 and 5 respectively. Further disclosed are nucleic acid sequences coding for these proteins (SEQ. ID NOS:2, 4 and 6).
  • Table 1 illustrates the homology (i.e. similarity) between the novel proteins DPRP-1, DPRP-2 and DPRP-3 and other known serine proteases.
  • Table 1 Comparison of the sequences of these three novel proteins with DPPIV and other Clan SC, Family S9 members and Subfamily B members
  • the expression pattern of DPRPs and the localization to specialized epithelial cells and plasma cells is consistent with a role in differentiation, proliferation and inflammation.
  • Data disclosed herein supports the hypothesis that DPRP-1 and DPRP-2 are involved in the regulation of proliferation of in vitro models of prostate and testis cancer well known to those skilled in the art.
  • DPRP-1 and DPRP-2 activities described herein and their expression patterns are compatible with their having functional roles as physiological regulators of the immune and neuroendocrine systems through the enzymatic modification of biochemical mediators like peptides and chemokines.
  • the numerous functions previously described for DPPIV based upon the use of inhibitors may be due in part to its action and that of similar proteins, like the DPRPs. Therefore, the discovery of selective and potent inhibitors of DPPIV, of the DPRPs and of other related proteases like FAP is considered central to achieving effective and safe pharmaceutical use of these and any newly identified serine protease inhibitors, as well as other active compounds that modify the function(s) of such proteins.
  • the invention thus provides novel proteins or polypeptides, the nucleic acids coding therefor, cells which have been modified with the nucleic acid so as to express these proteins, antibodies to these proteins, a screening method for the discovery of new therapeutic agents which are inhibitors of the activity of these proteins (or which are inhibitors of DPPIV and not of the proteins), and therapeutic agents discovered by such screening methods.
  • Tile novel proteins and the nucleic acids coding therefor can be used to discover new therapeutic agents for the treatment of certain diseases, such as for example, reproductive, inflammatory and metabolic disorders and also in the preparation of antibodies with therapeutic or diagnostic value.
  • novel, mature, biologically active proteins principally of human origin.
  • Such proteins may be isolated in small quantities from suitable animal (including human) tissue or biological fluids by standard techniques; however, larger quantities are more conveniently prepared in cultures of cells genetically modified so as to express the protein.
  • nucleic acid molecules encoding polypeptides of the present invention including mRNAs, DNAs, cDNAs, genomic DNAs thereof.
  • nucleic acid probes are also provided comprising nucleic acid molecules of sufficient length to specifically hybridize to a nucleic acid sequence of the present invention.
  • processes utilizing recombinant techniques are provided for producing such polypeptides useful for in vitro scientific research, for example, synthesis of DNA and manufacture of DNA vectors.
  • Processes for producing such polypeptides include culturing recombinant prokaryotic and/or eukaryotic host cells that have been transfected with DNA vectors containing a nucleic acid sequence encoding such a polypeptide and/or the mature protein under conditions promoting expression of such protein and subsequent recovery of such protein or a fragment of the expressed product.
  • the invention provides methods for using DPRP polypeptides and polynucleotides, including the treatment of infections, such as bacterial, fungal, protozoan and viral infections, particularly infections caused by HIV-1 or HIV-2, pain, diabetes, precocious puberty, infertility, obesity, anorexia, bulimia, Parkinson's disease, acute heart failure, hypotension, hypertension, urinary retention, osteoporosis, angina pectoris, myocardial infarction, stroke, ulcers, asthma, allergies, benign prostatic hypertrophy, cancers including hormone-sensitive and androgen- independent cancers, migraines, vomiting, psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, depression, dementia, and severe mental retardation, and dyskinesias, hereinafter collectively referred to as "the Diseases”.
  • infections such as bacterial, fungal, protozoan and viral infections, particularly infections caused by HIV-1 or HIV-2, pain, diabetes, precocious puberty, infertility, obesity, an
  • the invention provides iolated nucleic acid which encodes (a) a polypeptide which includes the amino acid sequence of one of SEQ ID NOS: 1, 3 and 5, or (b) a polypeptide having an amino acid sequence that is at least about 70% similar thereto and exhibits the same biological function, or which is an alternative splice variant of one of SEQ ID NOS:2, 4 and 6, or which is a probe comprising at least 14 contiguous nucleotides from said nucleic acid encoding (a) or (b), or which is complementary to any one of the foregoing.
  • the invention provides a polypeptide which may be optionally glycosylated, and which (a) has the amino acid sequence of a mature protein set forth in any one of SEQ ID NOS: 1, 3 and 5; (b) has the amino acid sequence of a mature protein having at least about 70% similarity to one of the mature proteins of (a) and which exhibits the same biological function; (c) has the amino acid sequence of a mature protein having at least about 90% identity with a mature protein of any of SEQ ID NOS:l, 3 and 5; or (d) is an immunologically reactive fragment of (a).
  • the invention provides a method for the screening for a compound capable of inhibiting the enzymatic activity of at least one mature protein of the invention, which method comprises incubating said mature protein and a suitable substrate for said mature protein in the presence of one or more test compounds or salts thereof, measuring the enzymatic activity of said mature protein, comparing said activity with comparable activity determined in the absence of a test compound, and selecting the test compound or compounds that reduce the enzymatic activity, and it also provides a method for screening for a compound capable of inhibiting the enzymatic activity of DPPIV that does not inhibit the enzymatic activity of at least one mature protein and a suitable substrate in the presence of one or more inhibitors of DPPIV or salts thereof, measuring the enzymatic activity of said mature protein, comparing said activity with comparable activity determined in the absence of the DPPIV inhibitor, and selecting a compound that does not reduce the enzymatic activity of said mature protein.
  • FIGS. 1A and IB show the co-linear alignment of DPRP-1, DPRP-2, DPRP-3 and DPPIV, with shading being supplied to indicate the same (black) or similar (gray) amino acid residues at a particular location.
  • FIG. 2 is similar to FIG. 1 and shows co-linear alignment of human and mouse
  • FIG. 3 is a graph which shows the effects of various tetrapeptide amide inhibitors on dipeptidyl peptidase enzyme activity.
  • FIGS. 4A-4C show the effects of three inhibitor compounds on the proliferation of PC3 prostate cancer cell lines at various doses.
  • isolated nucleic acid sequences which encode the mature polypeptides having the deduced amino acid sequences of the three DPRP's (SEQ ID NOS:l, 3 and 5).
  • the polynucleotides of this invention were discovered using a human testis cDNA library (DPRP-1), a human colon library (DPRP-2) and a human hypothalamus cDNA library (DPRP-3).
  • Isolated nucleic acid for DPRP-1 contains an open reading frame encoding a protein of approximately 882 amino acids in length which is structurally related to human DPPIV, showing 26% identity, and 41% similarity over the entire human DPPIV protein sequence.
  • Isolated nucleic acid for DPRP-2 contains an open reading frame encoding for a protein of approximately 864 amino acids, which is 39% similar to the entire DPPIV amino acid sequence.
  • Analysis of DPRP-1 and DPRP- 2 primary amino acid sequence using hydrophobicity plots predicts that these two proteins do not have a transmembrane domain. Despite this fact, it is possible that these intracellular serine proteases are secreted upon cellular activation.
  • QPP Quiescent cell proline dipeptidase
  • DPRP-1 and DPRP-2 serine protease that is targeted to intracellular vesicles that are distinct from lysosomes. This hypothesis expands the potential site(s) and scope of DPRP-1 and DPRP-2 involvement in mechanisms for post-translational regulation of chemokines, cytokines, peptides and polypeptides.
  • the full length DPRP-3 sequence contains 796 amino acids, a signal peptide from 1 to 48, and a transmembrane domain between 34 and 56.
  • the mature protein is predicted to be a type II membrane protein and may be cleaved to produce a soluble form.
  • the amino acid sequence is set forth in SEQ ID NO: 5 , which was deduced from SEQ ID NO:6 and has 54% similarity with DPPIV.
  • Amino acid sequence alignments of these polypeptides with members of the prolyloligopeptidase enzyme subfamily S9B show that all three DPRP proteins have overall sequence and structural homology to DPPIV and FAP.
  • DPRPs are predicted to be a members of the enzyme Clan SC (Serine nucleophile) with catalytic residues in the order Ser, Asp, His and the active site sequence (G-W-S-Y-G).
  • DPRP-1, DPRP-2 and DPRP-3 do not exhibit sequence similarity with any members of the classical serine protease families, chymotrypsin and subtilisin.
  • the order of the catalytic triad residues is different in the three main related SC clan families: His-
  • DPRP-3 has the highest homology with DPPVI (68% homology and 51% identity).
  • Wada et al isolated cDNA clones for DPPVI, a DPPIV- related protein, from bovine, rat (Wada et al., Proc. Nat. Acad. Sci. 89: 197-201. (1992)) and human (Yokotani et al., Hum. Molec. Genet. 2:1037-1039 (1993)) brain libraries.
  • DPPIV the catalytic triad in DPPVI does not have the first serine residue.
  • DPRP-3 two of the amino acids in the catalytic triad characteristic of the serine protease family are conserved. However, the serine residue itself is replaced by glycine. While the absence of the serine residue is likely to prevent protease activity at this site, it is possible that multiple other functions mediated by other functional domains of the protein remain intact.
  • DPPIV is a multifunctional molecule that exerts important functions depending on the expressed cells and tissues, in addition to its catalytic activity as a peptidase. DPRP-3 and DPPVI are also likely to maintain multiple functions despite the absence of an intact catalytic triad.
  • DPPVI has been implicated in the regulation of neuronal plasticity.
  • DPPVI is highly expressed in the hippocampus, thalamus, hypothalamus and stiatum.
  • developmental arrest and embryonic lethality of rump white Rw/Rw embryos is thought to be due to disruption of the DPPIV gene.
  • Rw mutation is associated with a chromosomal inversion spanning 30 cM of the proximal portion of mouse chromosome 5.
  • Genomic analysis of the DPPVI gene on the Rw chromosome places the inversion breakpoint in the coding region resulting in loss of a significant fraction of the C-terminal region, Hough R.B. et al., Proc. Nat. Acad. Sci.. 95. 13800-13805 (1998).
  • the human DPRP-1 gene predicted to be 32668bp in length, has at least 22 exons and eight transcripts. It maps to chromosome 15 (NT_010265) at position 15q21.1 - 15q22.1.
  • the lengths of predicted alternative splice variant transcripts vary between 602bp and 4523bp (see SEQ ID NOS: 7-22). This is in agreement with the multiple transcripts observed by Northern blot analysis (See Example 2).
  • Human DPRP-2 belongs to a gene with at least 27 exons and nine splice variants
  • the DPRP-2 gene maps to region 19pl3.3 of chromosome 19. This location is host to a number of disease markers and is associated with various disorders including hypocalciuric hypercalcemia, type II cerebellar ataxia, muscular dystrophy, convulsions, susceptibility to atherosclerosis, psoriasis, ectodermal dysplasia, and acute myeloid leukemia. In agreement with the ubiquitous distribution of the mRNA observed by Northern blot analysis (see Example 2), DPRP-2 was expressed in a wide variety of tissues upon examination of EST's coverage (e.g.
  • Human DPRP-3 belongs to a gene with at least 23 exons and two splice variants (see SEQ ID NOS:41-44). The gene maps to chromosome 2 (NT_005445) at position 2ql2.3-2ql4.1. Transcripts for DPRP-3 did not show as wide a distribution as DPRP-1 and DPRP-2. As shown by Northern blot in Example 2, DPRP-3 expression is restricted to brain and pancreas. ESTs representing the DPRP-3 mRNA were abundant in tissue derived from multiple sclerosis lesions, hypothalamus, whole brain and nerves, with a few transcripts being found in uterus and colon.
  • the polynucleotides of the present invention may be in the form of RNA or in the form of DNA; DNA should be understood to include cDNA, genomic DNA, and synthetic DNA.
  • the DNA may be double-stranded or single-stranded and, if single- stranded, may be the coding strand or non-coding (antisense) strand.
  • the coding sequence which encodes the mature polypeptide may be identical to the coding sequence shown in SEQ ID NOS:2, 4 and 6 respectively, or it may be a different coding sequence encoding the same mature polypeptide, as a result of the redundancy or degeneracy of the genetic code or a single nucleotide polymorphism.
  • RNA transcript which includes the entire length of any one of SEQ ID NOS:2, 4 and 6.
  • the polynucleotides which encode the mature proteins of SEQ ID NOS:l, 3, 5, respectively, may include but are not limited to the coding sequence for the mature protein alone; the coding sequence for the mature polypeptide plus additional coding sequence, such as a leader or secretory sequence or a proprotein sequence; and the coding sequence for the mature protein (and optionally additional coding sequence) plus non-coding sequence, such as introns or a non-coding sequence 5' and/or 3' of the coding sequence for the mature protein.
  • polynucleotide encoding a polypeptide or the term “nucleic acid encoding a polypeptide” should be understood to encompass a polynucleotide or nucleic acid which includes only coding sequence for the mature protein as well as one which includes additional coding and/or non-coding sequence.
  • polynucleotides and nucleic acid are used interchangeably.
  • the present invention also includes polynucleotides where the coding sequence for the mature protein may be fused in the same reading frame to a polynucleotide sequence which aids in expression and secretion of a polypeptide from a host cell; for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell may be so fused.
  • the polypeptide having such a leader sequence is termed a preprotein or a preproprotein and may have the leader sequence cleaved, by the host cell to form the mature form of the protein.
  • These polynucleotides may have a 5' extended region so that it encodes a proprotein, which is the mature protein plus additional amino acid residues at the N-terminus.
  • polynucleotides of the present invention may encode mature proteins, or proteins having a prosequence, or proteins having both a prosequence and a presequence (leader sequence).
  • the polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptides of the present invention.
  • the marker sequence may be a polyhistidine tag, a hemagglutinin (HA) tag, a c-myc tag or a V5 tag when a mammalian host, e.g. COS-1 cells, is used.
  • the HA tag would correspond to an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al., Cell 37:767 (1984)), and the c-myc tag may be an eptitope from human Myc protein (Evans, G.I. et al., Mol. Cell. Biol 5: 3610-3616 (1985)).
  • gene means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons).
  • significant sequence homology is intended to denote that at least 25%, preferably at least 40%, of the amino acid residues are conserved, and that, of the non- conserved residues, at least 40% are conservative substitutions. Fragments of the full-length genes of the present invention may be used as a hybridization probe for a cDNA library to isolate full-length cDNA as well as to isolate other cDNAs which have significant sequence homology to the gene and will encode proteins or polypeptides having similar biological activity or function.
  • Such a probe of this type has at least 14 bases (at least 14 contiguous nucleotides from one of SEQ ID NOS:2, 4 or 6), preferably at least 30 bases, and such may contain, for example, 50 or more bases.
  • Such probe may also be used to identify a cDNA clone corresponding to a full-length transcript and/or a genomic clone or clones that contains the complete gene, including regulatory and promoter regions, exons, and introns.
  • Labelled oligonucleotides having a sequence complementary to that of the gene of the present invention are useful to screen a library of human cDNA, genomic DNA or mRNA to locate members of the library to which the probe hybridizes.
  • a known DNA sequence may be used to synthesize an oligonucleotide probe which is then used in screening a library to isolate the coding region of a gene of interest.
  • the present invention is considered to further provide polynucleotides which hybridize to the hereinabove-described sequences wherein there is at least 70%, preferably at least 90%, and more preferably at least 95% identity or similarity between the sequences, and thus encode proteins having similar biological activity.
  • identity between two polypeptides when the amino acid sequences contain the same or conserved amino acid substitutes for each individual residue in the sequence. Identity and similarity may be measured using sequence analysis software (e.g., Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, WI 53705).
  • stringent conditions means conditions which permit hybridization between polynucleotides sequences and the polynucleotide sequences of SEQ ID NOS. -2, 4 and 6 where there is at least about 70% identity.
  • stringent conditions can be defined by, e.g., the concentrations of salt or formamide in the prehybridization and hybridization solutions, or by the hybridization temperature, and are well known in the art.
  • stringency can be increased by reducing the concentration of salt, by increasing the concentration of formamide, and/or by raising the hybridization temperature.
  • hybridization under high stringency conditions may employ about
  • hybridization under reduced stringency conditions might employ about 35% to 25% formamide at about 30°C to 35°C.
  • One particular set of conditions for hybridization under high stringency conditions employs 42°C, 50% formamide, 5x. SSPE, 0.3% SDS, and 200 ⁇ g/ml sheared and denatured salmon sperm DNA.
  • similar conditions as described above may be used in 35% formamide at a reduced temperature of 35°C.
  • the temperature range corresponding to a particular level of stringency can be further narrowed by calculating the purine to pyrimidine ratio of the nucleic acid of interest and adjusting the temperature accordingly. Variations on the above ranges and conditions are well known in the art.
  • hybridization should occur only if there is at least 95%o, and more preferably at least 97%, identity between the sequences.
  • the polynucleotides which hybridize to the hereinabove described polynucleotides in a preferred embodiment encode polypeptides which exhibit substantially the same biological function or activity as the mature protein encoded by one of the cDNAs of SEQ ID NOS:2, 4 and 6.
  • a suitable polynucleotide probe may have at least 14 bases, preferably 30 bases, and more preferably at least 50 bases, and will hybridize to a polynucleotide of the present invention which has an identity thereto, as hereinabove described, and which may or may not retain activity.
  • such polynucleotides may be employed as a probe for hybridizing to the polynucleotides of SEQ ID NOS:2, 4 and 6 respectively, for example, for recovery of such a polynucleotide, or as a diagnostic probe, or as a PCR primer.
  • the present invention includes polynucleotides having at least a 70% identity, preferably at least a 90% identity, and more preferably at least a 95%» identity to a polynucleotide which encodes the polypeptides of SEQ ID NOS: 1, 3 and 5 respectively, as well as fragments thereof, which fragments preferably have at least 30 bases and more preferably at least 50 bases, and to polypeptides encoded by such- polynucleotides .
  • the genetic code is redundant in that certain amino acids are coded for by more than one nucleotide triplet (codon), and the invention includes those polynucleotide sequences which encode the same amino acids using a different codon from that specifically exemplified in the sequences herein.
  • Such a polynucleotide sequence is referred to herein as an "equivalent" polynucleotide sequence.
  • the present invention further includes variants of the hereinabove described polynucleotides which encode for fragments, such as part or all of the mature protein, analogs and derivatives of one of the polypeptides having the deduced amino acid sequence of SEQ ID NOS:l, 3 and 5 respectively.
  • the variant forms of the polynucleotides may be a naturally occurring allelic variant of the polynucleotides or a non-naturally occurring variant of the polynucleotides.
  • the variant in the nucleic acid may simply be a difference in codon sequence for the amino acid resulting from the degeneracy of the genetic code, or there may be deletion variants, substitution variants and addition or insertion variants.
  • an allelic variant is an alternative form of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides that does not substantially alter the biological function of the encoded polypeptide.
  • the present invention further includes polypeptides which have the deduced amino acid sequence of SEQ ID NOS:l, 3 and 5, as well as fragments, analogs and derivatives of such polypeptides.
  • fragment when referring to the polypeptides of SEQ ID NOS:l, 3 and 5, means polypeptides that retain essentially the same biological function or activity as such polypeptides.
  • An analog might, for example, include a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature protein.
  • polypeptides of the present invention may be recombinant polypeptides, natural polypeptides or synthetic polypeptide; however, they are preferably recombinant polypeptides, glycosylated or unglycosylated.
  • the fragment, derivative or analog of a polypeptide of SEQ ID NOS: 1, 3 and 5 respectively may be (i) one in which one or more of the amino acid residues is substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which additional amino acids are fused to the mature protein, such as a leader or secretory sequence or a sequence which is employed for purification of the mature polypeptide or a proprotein sequence.
  • polypeptides and polynucleotides of the present invention should be in an . isolated form, and preferably they are purified to substantial homogeneity or purity.
  • substantial homogeneity is meant a purity of at least about 85%.
  • isolated is used to mean that the material has been removed from its original environment (e.g., the natural environment if it is naturally occurring).
  • a naturally occurring polynucleotide or polypeptide present in a living animal is not considered to be isolated, but the same polynucleotide or polypeptide, when separated from substantially all of the coexisting materials in the natural system, is considered isolated.
  • the term includes, for example, a recombinant DNA which is incorporated into a vector, into an autonomously replicating plasmid or virus, or into the genomic DNA of a prokaryote or eukaryote; or which exists as a separate molecule (e.g., a cDNA or a genomic or cDNA fragment produced by polymerase chain reaction (PCR) or restriction endonuclease digestion) independent of other sequences. It also includes a recombinant DNA which is part of a hybrid gene encoding additional polypeptide sequence, e.g., a fusion protein.
  • polypeptides of the present invention include any one of the polypeptide of
  • SEQ ID NOS:l, 3 and 5 in particular the mature protein
  • polypeptides which have at least 70% similarity e.g. preferably at least 60% and more preferably at least 70% identity
  • polypeptides of SEQ ID NOS:l, 3 and 5 more preferably at least 90% similarity (e.g. preferably at least 90% identity) to one of the polypeptides of SEQ ID NOS:l, 3 and 5, and most preferably at least 95% similarity (e.g. preferably at least 95% identity) to one of the polypeptides of SEQ ID NOS:l, 3 and 5.
  • Fragments or portions of the polypeptides of the present invention may be employed as intermediates for producing the corresponding full-length polypeptides by peptide synthesis. Fragments or portions of the polynucleotides of the present invention may also be used to synthesize full-length polynucleotides of the present invention.
  • the present invention also includes vectors which include such polynucleotides, host cells which are genetically engineered with such vectors and the production of polypeptides by recombinant techniques using the foregoing.
  • Host cells are genetically engineered (transduced or transformed or transfected) with such vectors which may be, for example, a cloning vector or an expression vector.
  • the vector may be, for example, in the form of a plasmid, a viral particle, a phage, etc.
  • the engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the genes of the present invention.
  • the culture conditions such as temperature, pH and the like, are those commonly used with the host cell selected for expression, as well known to the ordinarily skilled artisan.
  • the polynucleotides of the present invention may be employed for producing polypeptides by recombinant techniques.
  • the polynucleotides may be included in any one of a variety of expression vectors for expressing polypeptides.
  • Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, and pseudorabies.
  • any other vector may be used as long as it is replicable and viable in the host.
  • the appropriate DNA sequence may be inserted into the vector by any of a variety of procedures.
  • the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by procedures well known in the art, which procedures are deemed, to be within the scope of those skilled in this art.
  • the DNA sequence in the expression vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis.
  • promoters there may be mentioned: LTR or S V40 promoter, the E. coli. lac or trp, the phage lambda P.sub.L promoter and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses.
  • the expression vector should also contain a ribosome binding site for translation initiation and a transcription terminator.
  • the vector may also include appropriate sequences for amplifying expression.
  • the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells, such as dihydrofolate reductase or neomycin-resistance for eukaryotic cell culture, or such as tetracycline- or ampicillin-resistance in E. coli.
  • the vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the protein.
  • appropriate hosts there may be mentioned: bacterial cells, such as E. coli, Streptomyces, Salmonella typhimurium; fungal cells, such as yeast; insect cells, such as Drosophila S2 and Spodoptera Sf9; animal cells, such as CHO, COS or Bowes melanoma; adenoviruses; plant cells, etc.
  • bacterial cells such as E. coli, Streptomyces, Salmonella typhimurium
  • fungal cells such as yeast
  • insect cells such as Drosophila S2 and Spodoptera Sf9
  • animal cells such as CHO, COS or Bowes melanoma
  • adenoviruses adenoviruses
  • plant cells etc.
  • the selection of an appropriate host is deemed to be within the scope of those skilled in the
  • the present invention further includes recombinant constructs comprising one or more of the sequences as broadly described above.
  • the constructs may comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation.
  • the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence.
  • a promoter operably linked to the sequence.
  • Bacterial pQE70, pQE60, pQE-9 (Qiagen), pBS, pDIO, phagescript, psiX174, pbluescript SK, pbsks, pNH8A, pNH16a, pNHl ⁇ A, pNH46A (Stratagene), ptrc99a, pK 223-3, p K233-3, pDR540 and pRIT5 (Pharmacia); and Eukaryotic: pWLNEO, pSV2CAT, pOG44, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, and pSVL (Pharmacia).
  • any other suitable plasmid or vector may be used as long as it is replicable and viable in the host.
  • Promoter regions can be selected from any desired gene using CAT (chloramphenicol acetyl transferase) vectors or other vectors with selectable markers.
  • Two appropriate vectors are pKK232-8 and pCM7.
  • Particular named bacterial promoters include lad, lacZ, T3, T7, gpt, lambda P.sub.R, P.sub.L and trp.
  • Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
  • Components of the expression vector may generally include: 1) a neomycin phosphotransferase (G418), or hygromycin B phosphotransferase (hyg) gene as a selection marker, 2) an E. coli origin of replication, 3) a T7 and SP6 phage promoter sequence, 4) lac operator sequences, 5) the lactose operon repressor gene (laclq) and 6) a multiple cloning site linker region.
  • G408 neomycin phosphotransferase
  • hyg hygromycin B phosphotransferase
  • laclq lactose operon repressor gene
  • a multiple cloning site linker region Such an origin of replication (oriC) may be derived from pUC19 (LTI, Gaithersburg, Md.).
  • a nucleotide sequence encoding one of the polypeptides SEQ ID NOS:2,4 and 6 having the appropriate restriction sites is generated, for example, according to the PCR protocol described in Example 1 hereinafter, using PCR primers having restriction sites for Kpnl (as the 5' primer) and Notl or Sad (as the 3' primer) for DPRP-1, or sites for Hindlll (as the 5' primer) and Notl or BamHI (as the 3 ' primer) for DPRP-2.
  • the PCR inserts are gel-purified and digested with compatible restriction enzymes.
  • the insert and vector are ligated according to standard protocols.
  • the present invention provides host cells containing the above-described constructs.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, lipofection or electroporation (Davis, L., Dibner, M., Battey, I., Basic Methods in Molecular Biology, (1986)).
  • polypeptides of the invention can be synthetically produced by conventional peptide synthesizers or by chemical ligation of suitable fragments thus prepared.
  • Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989).
  • Enhancers include cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription. Examples include the S V40 enhancer on the late side of the replication origin bp 100 to 270, a cytomegalo virus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin- resistance gene of E. coli and S.
  • heterologous structural sequence is assembled in appropriate phase ' with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium.
  • the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
  • Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter.
  • the vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desired, provide amplification within the host.
  • Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
  • useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017).
  • cloning vector pBR322 ATCC 37017
  • Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and G ⁇ M1 (Promega Biotec, Madison, Wis., U.S.A.). These pBR322 "backbone" sections are combined with an appropriate promoter and the structural sequence to be expressed.
  • the selected promoter is induced by appropriate means (e.g., temperature shift or chemical induction), and cells are cultured for an additional period.
  • Cells are typically harvested by centrifugation and then disrupted by physical or chemical means, with the resulting crude extract being retained for further purification.
  • Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption and use of cell-lysing agents; such methods are well known to those skilled in the art.
  • mammalian cell culture systems can also be employed to express a recombinant protein.
  • mammalian expression systems include the COS-7 lines of monkey kidney f ⁇ broblasts, described by Gluzman, Cell. 23:175 (1981).
  • Other cell lines capable of expressing a compatible vector include, for example, the C127, 3T3, CHO, HeLa and BHK cell lines.
  • Mammalian expression vectors will generally comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5' flanking nontranscribed sequences. DNA sequences derived from the SV40 splice, and polyadenylation sites may be used to provide required nontranscribed genetic elements.
  • the polypeptides can be recovered and purified from recombinant cell cultures by methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Recovery can be facilitated if the polypeptide is expressed at the surface of the cells, but such is not a prerequisite. Recovery may also be desirable of cleavage products that are cleaved following expression of a longer form of the polypeptide. Protein refolding steps as known in this art can be used, as necessary, to complete configuration of the mature protein. High performance liquid chromatography (HPLC) can be employed for final purification steps.
  • HPLC high performance liquid chromatography
  • polypeptides of the present invention may be purified natural products, or produced by recombinant techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect or mammalian cells in culture). Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. Polypeptides of the invention may also include an initial methionine amino acid residue.
  • the proteins of the invention are isolated and purified so as to be substantially free of contamination from other proteins.
  • the proteins of the invention should constitute at least 80% by weight of the total protein present in a sample, more preferably at least 90%, even more preferably at least 95%, and most preferably at least 98% by weight of the total protein.
  • proteins may be in the form of a solution in water, another suitable solvent, such as dimethyl sulphoxide (DMSO) or ethanol, or a mixture of suitable solvents.
  • suitable solvent such as dimethyl sulphoxide (DMSO) or ethanol
  • mixtures of solvents include 10% (by weight) ethanol in water and 2% (by weight) DMSO in water.
  • a solution may further comprise salts, buffering agents, chaotropic agents, detergents, preservatives and the like.
  • the proteins may be in the form of a solid, such as a lyophilised powder or a crystalline solid, which may also comprise a residual solvent, a salt or the like.
  • antibodies includes polyclonal antibodies, affinity-purified polyclonal antibodies, monoclonal antibodies, and antigen-binding fragments, such as F(ab') 2 and Fab' proteolytic fragments. Genetically engineered intact antibodies or fragments, such as chimeric antibodies, Fv fragments, single chain antibodies and the like, as well as synthetic antigen-binding peptides and polypeptides, are also included.
  • Non-human antibodies may be humanized by grafting non-human CDRs onto human framework and constant regions, or by incorporating the entire non-human variable domains (optionally "cloaking" them with a human-like surface by replacement of exposed residues, wherein the result is a "veneered” antibody). In some instances, humanized antibodies may retain non-human residues within the human variable region framework domains to enhance proper binding characteristics. Through humanizing antibodies, biological half-life may be increased, and the potential for adverse immune reactions upon administration to humans should be reduced.
  • Alternative techniques for generating or selecting antibodies useful herein include in vitro exposure of lymphocytes to human prohormone DPRP protein or a peptide therefrom, and selection of antibody display libraries in phage or similar vectors (for instance, through use of immobilized or labeled human DPRP protein or peptide).
  • Genes encoding polypeptides having potential human DPRP polypeptide binding domains can be obtained by screening random peptide libraries displayed on phage (phage display) or on bacteria, such as E. coli. Nucleotide sequences encoding such polypeptides can be obtained in a number of ways well known in this art.
  • polyclonal antibodies can be generated from inoculating a variety of warm-blooded animals, such as horses, cows, goats, sheep, dogs, chickens, rabbits, mice and rats, with a human DPRP polypeptide or a fragment thereof.
  • the immunogenicity of a human prohormone DPRP polypeptide may be increased through the use of an adjuvant, such as alum (aluminum hydroxide) or Freund's complete or incomplete adjuvant, or surface active substances, such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, KLH or dinitrophenol.
  • an adjuvant such as alum (aluminum hydroxide) or Freund's complete or incomplete adjuvant
  • surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, KLH or dinitrophenol.
  • Polypeptides useful for immunization also include fusion polypeptides, such as fusions of DPRP or a portion thereof with an immunoglobulin polypeptide or with maltose binding protein.
  • the polypeptide immunogen may be a full-length molecule or a portion thereof. If the polypeptide portion is "hapten-like", such portion may be advantageously joined or linked to a macromolecular carrier, such as keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA) or tetanus toxoid, for immunization.
  • KLH keyhole limpet hemocyanin
  • BSA bovine serum albumin
  • Antibodies to DPRP may also be generated using methods that are well known in the art. Such antibodies may include, but are not limited to, polyclonal, monoclonal, chimeric, and single chain antibodies, Fab fragments, and fragments produced by a Fab expression library. Neutralizing antibodies (i.e., those which block or modify interactions at the active sites) are especially preferred for therapeutic use.
  • libraries of single chain antibodies, Fab fragments, other antibody fragments, non-antibody protein domains, or peptides may be screened.
  • the libraries could be generated using phage display, other recombinant DNA methods, or peptide synthesis (Vaughan, T. J. et al. Nature Biotechnology 14: 309-314 (1966)). Such libraries would commonly be screened using methods which are well known in the art to identify sequences which demonstrate specific binding to DPRP.
  • the oligopeptides, peptides, or fragments used to induce antibodies to DPRP have an amino acid sequence consisting of at least about 5 amino acids and, more preferably, of at least about 10 amino acids. It is also preferable that these oligopeptides, peptides, or fragments are identical to a portion of the amino acid sequence of the natural protein. Short stretches of DPRP amino acids may also be fused with those of another protein, such as KLH, and antibodies to the chimeric molecule may be produced.
  • Monoclonal antibodies to DPRP may be prepared using any well known technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique, although monoclonal antibodies produced by hybridoma cells may be preferred.
  • chimeric antibodies such as the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used, see Neuberger, M.S. et al. Nature 312: 604-608 (1984).
  • techniques described for the production of single chain antibodies may be adapted, using methods known in the art, to produce DPRP-specific single chain antibodies.
  • Antibodies with related specificity, but of distinct idiotypic composition may be generated by chain shuffling from random combinatorial immunoglobulin libraries. (Burton D. R. Proc. Natl. Acad. Sci. 88: 11120-11123 (1991)).
  • Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature. (Orlandi, R. et al. Proc. Natl. Acad. Sci. 86: 3833-3837 (1989)).
  • Antibody fragments which contain specific binding sites for DPRP may also be generated.
  • fragments include, but are not limited to, F(ab') 2 fragments produced by pepsin digestion of the antibody molecule and Fab fragments generated by reducing the disulf ⁇ de bridges of the F(ab') 2 fragments.
  • Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity. (Huse, W. D. et al. Science 254: 1275-1281 (1989)).
  • Various immunoassays may be used to identify antibodies having the desired specificity.
  • compositions suitable for use in this aspect of the invention include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose relating to one of the Diseases.
  • the determination of a therapeutically effective dose is well within the capability of those skilled in the art and can be estimated initially either in cell culture assays, e.g. of neoplastic cells, or in animal models, usually mice, rats, rabbits, dogs, or pigs.
  • An animal model may also be used to determine the appropriate concentration range and route of administration, which information is then commonly used to determine useful doses and routes for administration in humans.
  • a therapeutically effective dose refers to that amount of active ingredient, e.g. a DPRP or fragment thereof, antibodies of DPRP, or an agonist, antagonist or inhibitor of DPRP, which ameliorates particular symptoms or conditions of the Disease.
  • the amount to be administered may be effective to cleave a desired target substrate upon contact therewith.
  • Therapeutic efficacy and toxicity may likewise be determined by standard pharmaceutical procedures in cell cultures or with experimental animals, such as by calculating the ED50 (the dose therapeutically effective in 50% of the population) or LD50 (the dose lethal to 50% of the population) statistics.
  • the dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the LD50/ED50 ratio.
  • Pharmaceutical compositions which exhibit large therapeutic indices are preferred.
  • the data obtained from cell culture assays and animal studies is used in formulating a range of dosage for human use.
  • the dosage contained in such compositions is preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage varies within this range depending upon the dosage form employed, the sensitivity of the patient, and the route of administration.
  • An exact dosage will normally be determined by the medical practitioner in light of factors related to the subject requiring treatment, with dosage and administration being adjusted to provide a sufficient level of the active moiety or to maintain a desired effect. Factors to be taken into account include the severity of the disease state, the general health of the subject, the age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical compositions may be administered every 3 to 4 days, every week, or even once every two weeks, depending on the half-life and clearance rate of the particular formulation.
  • Yet another aspect of the invention provides polynucleotide molecules having sequences that are antisense to mRNA transcripts of DPRP 1, DPRP2 and DPRP-3 polynucleotides.
  • Administration of an antisense polynucleotide molecule can block the production of the protein encoded by DPRP-1, DPRP2 or DPRP-3.
  • the techniques for preparing antisense polynucleotide molecules and administering such molecules are known in the art.
  • antisense polynucleotide molecules can be encapsulated into liposomes for fusion with cells.
  • the expression of DPRP-1, DPRP-2 and DPRP-3 in specialized epithelial cells, immune cells (lymphocytes and B cells), astrocytic tumors, and in various hormone sensitive cancers provides evidence of a potential role in the pathophysiology of cancer, metaplasia and metastasis. Therefore in a further aspect, the invention relates to diagnostic assays for detecting diseases associated with inappropriate DPRP activity or expression levels. Antibodies that specifically bind DPRP may be used for the diagnosis of disorders characterized by expression of DPRP, or in assays to monitor patients being treated with DPRP or with agonists or antagonists (inhibitors) of DPRP.
  • Antibodies useful for diagnostic purposes may be prepared in the same manner as those described above for therapeutics. Diagnostic assays for DPRP include methods that utilize the antibody and a label to detect DPRP in human body fluids or in extracts of cells or tissues.
  • the antibodies may be used with or without modification, and they may be labeled by covalent or non-covalent joining with a reporter molecule.
  • reporter molecules A wide variety of reporter molecules are known in the art.
  • Recombinant DPRP proteins that have been modified so as to be catalytically inactive can also be used as dominant negative inhibitors. Such modifications include, for example, mutation of the active site.
  • a variety of protocols for measuring DPRP including ELISAs, RIAs and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of DPRP expression.
  • Normal or standard values for DPRP expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, preferably human, with antibody to DPRP under conditions suitable for complex formation.
  • the method for detecting DPRP in a biological sample would comprise the steps of: a) providing a biological sample; b) combining the biological sample and an anti-DPRP antibody under conditions which are suitable for complex formation to occur between DPRP and the antibody; and c) detecting complex formation between DPRP and the antibody, thereby establishing the presence of DPRP in the biological sample.
  • the amount of complex formation then may be quantified by various methods, preferably by photometric means. Quantities of DPRP expressed in subject, control, and disease samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease.
  • the polynucleotides encoding DPRP are used for diagnostic purposes, which polynucleotides may include oligonucleotide sequences, complementary RNA and DNA molecules, and PNAs. These polynucleotides may be used to detect and quantitate gene expression in biopsied tissues in which expression of DPRP may be correlated with one of the Diseases.
  • the diagnostic assay may be used to distinguish between absence, presence, and excess expression of DPRP and to monitor regulation of DPRP levels during therapeutic intervention.
  • pharmacogenomic, single nucleotide polymorphisms (SNP) analysis of the DPRP genes can be used as a method to screen for mutations that indicate predisposition to disease or modified response to drugs.
  • DPRP polynucleotide and polypeptide sequences, fragments thereof, antibodies of DPRPs, and agonists, antagonists or inhibitors of DPRPs can be used to as discovery tools to identify molecular recognition events and therefore proteins, polypeptides and peptides that interact with DPRP proteins.
  • a specific example is phage display peptide libraries where greater than 108 peptide sequences can be screened in a single round of panning.
  • Such methods as well as others are known within the art and can be utilized to identify compounds that inhibit or enhance DPRP-1, DPRP-2 or DPRP-3 activity.
  • Coupled links represent functional interactions such as complexes or pathways, and proteins that interact with DPRPs can be identified by a yeast two-hybrid system, proteomics (differential 2D gel analysis and mass spectrometry) and genomics (differential gene expression by microarray or serial analysis of gene expression SAGE). Proteins identified as functionally linked to DPRPs and the process of interaction form the basis of methods of screening for inhibitors, agonists and antagonists and modulators of these DPRP-protein interactions.
  • Antagonist refers to an inhibitor molecule which, when bound to DPRP, decreases the amount or the duration of the effect of the biological or immunological activity of DPRP, e.g. decreasing the enzymatic activity of the peptidase to cleave the N-terminal dipeptide.
  • Antagonists may include proteins, nucleic acids, carbohydrates, antibodies, or any other molecules which decrease the effect of DPRP; for example, they may include small molecules and organic compounds that bind to and inactivate DPRPs by a competitive or non-competitive type mechanism. Specific examples of DPRP tetrapeptide peptidic enzyme activity inhibitors are described in Example 6 and 7.
  • Inhibitors can be, for example, inhibitors of the DPRP protease activity, or alternatively inhibitors of the binding activity of the DPRP to proteins with which they interact.
  • Specific examples of such inhibitors can include, for example, anti-DPRP antibodies, peptides, protein fragments, or small peptidyl protease inhibitors, or small non-peptide, organic molecule inhibitors which are formulated in a medium that allows introduction into the desired cell type.
  • such inhibitors can be attached to targeting ligands for introduction by cell-mediated endocytosis and other receptor mediated events. Such methods are described further below and can be practiced by those skilled in the art given the DPRP nucleotide and amino acid sequences described herein.
  • a further use for DPRPs is for the screening of potential antagonists for use as therapeutic agents, for example, for inhibiting binding to DPRP, as well as for screening for agonists.
  • DPRP, its immunogenic fragments, or oligopeptides thereof can be used for screening libraries of compounds which are prospective agonists or antagonists in any of a variety of drug screening techniques.
  • the fragment employed in such screening may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly. The formation of binding complexes between DPRP and the agent being tested is then measured.
  • Other assays to discover antagonists that will inhibit DPRP are apparent from the disclosures of U.S. Patents Nos.
  • a method provided for screening a library of small molecules to identify a molecule which binds DPRP generally comprises: a) providing a library of small molecules; b) combining the library of small molecules with the polypeptide of either SEQ ID NOS: 1, 3 or 5, or with a fragment thereof, under conditions which are suitable for complex formation; and c) detecting complex formation, wherein the presence of such a complex identifies a, small molecule which binds DPRP.
  • One method for identifying an antagonist comprises delivering a small molecule which binds DPRP into extracts from cells transformed with a vector expressing DPRP along with a chromogenic substrate (e.g. Ala-Pro-AFC or Ala-Pro-AMC) under conditions where cleavage would normally occur, and then assaying for inhibition of cleavage by the enzyme by monitoring changes in fluorescence, or UV light absorption, by spectrophotometry to identify molecules that inhibit cleavage.
  • a reduced rate of reaction or total amount of fluorescence or UV light absorption, in the presence of the molecule establishes that the small molecule is an antagonist which reduces DPRP catalytic/enzymatic activity. Once such molecules are identified, they may be administered to reduce or inhibit cleaving by a DPRP.
  • agonist refers to a molecule which, when bound to DPRP, increases or prolongs the duration of the effect of DPRP.
  • Agonists may include proteins, nucleic acids, carbohydrates, or any other molecules that bind to and modulate the effect of DPRP.
  • a method for identifying such a small molecule, which binds DPRP as an agonist comprises delivering a chromogenic form of a small molecule that binds DPRP into cells transformed with a vector expressing DPRP and assaying for fluorescence or UV light absorption changes by spectrophotometry. An increased amount of UV absorption or fluorescence would establish that the small molecule is an agonist that increases DPRP activity.
  • Another technique for drug screening provides for high throughput screening of compounds having suitable binding affinity to the protein of interest as described in published PCT application WO 84/03564.
  • large numbers of different small test compounds are synthesized on a solid substrate, such as plastic pins or some other surface.
  • the test compounds are reacted with DPRP, or with fragments thereof, and then washed.
  • Bound DPRP is then detected by methods well known in the art.
  • Purified DPRP can also be coated directly onto plates for use in the aforementioned drug screening techniques.
  • non-neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support.
  • ligands may be designed that, for example, have more interactions with DPRP than do its natural ligands. Such antagonist ligands will bind to DPRP with higher affinity and so function as competitive ligands.
  • synthetic or recombinant proteins homologous or analogous to the ligand binding site of native DPRP may be designed, as may other molecules having high affinity for DPRP. Such molecules should also be capable of displacing DPRP and provide a protective effect.
  • the invention provides such cell lines engineered with Lox-Neo IRES tk cassette and GFP-IRES-Neo Knock-in/out cassette DNA element for constructing somatic gene targeting vectors.
  • Example 1 Cloning and Expression of DPRP genes Using the Mammalian Expression System DNA fragments encoding the full-length polypeptide DPRP-1 were amplified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of the gene, i.e. SEQ ID NO:45 and NO:46.
  • DNA fragments encoding the full length polypeptide DPRP-2 were amplified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of that gene, i.e. SEQ ID NO:50 and NO:51.
  • DNA fragments encoding the full length polypeptide DPRP-3 were amplified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of that gene, i.e. SEQ ID NO:55 and NO:56.
  • the three amplified sequences were respectively isolated from a 0.7% agarose gel using commercially available kit (GFX PCR DNA and Gel Band Purification Kit, Amersham Pharmacia Biotech Inc., Piscataway NJ, USA). The fragments were then ligated into cloning vector, pGEM-7Zf(-) (Promega Corporation, Madison WI, USA) and sequenced.
  • the corresponding cloning constructs were respectively designated ⁇ GEM7-DPRPl, pGEM7-DPRP2 and pGEM7-DPRP3.
  • the DNA sequences encoding the truncated DPRP-1 or DPRP-2 or DPRP-3 were amplified using pGEM7-DPRPl or pGEM7-DPRP2 or pGEM7-DPRP3 as a template and PCR oligonucleotide primers.
  • SEQ ID NO:45 and NO:47 were used for DPRP-1;
  • SEQ ID NO:50 and NO:52 were used for DPRP-2; and
  • SEQ ID NO:57 and NO:58 for DPRP-3.
  • the amplified sequences were again isolated from a 0.7% agarose gel using the same purification kits and sub-cloned into pGEM-7Zf(-).
  • the resulting constructs were designated pGEM7-DPRPlf, pGEM7- DPRP2f and pGEM7-DPRP3f.
  • pGEM7-DPRPl was digested with the restriction enzymes Kpnl and Notl to release the full length DPRP-1 gene.
  • the DNA fragment carrying the DPRP-1 gene was gel band purified using the above kit and then inserted into expression vector pcDNA3 (Invitrogen, Carlsbad CA, USA) to make the native DPRP-1 expression construct, which was designated pcDNA- DPRP1.
  • pGEM7-DPRPlf was digested with the restriction enzymes Xbal and Hindlll to release the truncated DPRP- 1 f gene.
  • the DNA fragment carrying the DPRP- 1 f gene was gel band purified using the above kit and then inserted into expression vector pcDNA3.1(-)/myc-His A (Invitrogen, Carlsbad CA, USA) to make the tagged DPRP-1 expression construct pcDNA-MycHis-DPRPl.
  • pGEM7-DPRP2 was digested with the restriction enzymes Hindlll and BamHI to release the full length DPRP-2 gene.
  • the DNA fragment carrying the DPRP-2 gene was gel band purified using the above kit and then inserted into expression vector pcDNA3 (Invitrogen, Carlsbad CA, USA) to make the native DPRP-2 expression construct, which was designated pcDNA-DPRP2.
  • pGEM7-DPRP2f was digested with the restriction enzymes EcoRI and BamHI to release the truncated DPRP-2f gene.
  • the DNA fragment carrying the DPRP-2f gene was gel band purified using the above kit and then inserted into expression vector pcDNA3.1(-)/myc-His B (Invitrogen, Carlsbad CA, USA) to make the tagged DPRP-2 expression construct designated pcDNA-MycHis-DPRP2.
  • pGEM7-DPRP3 was digested with the restriction enzymes EcoRI and Xhol to release the full length DPRP-3 gene.
  • the DNA fragment carrying the DPRP-3 gene was gel band purified using the above kit and then inserted into expression vector pcDNA3 (Invitrogen, Carlsbad CA, USA) to make the native DPRP-3 expression construct designated pcDNA-DPRP3.
  • pGEM7-DPRP3f was digested with the restriction enzymes Nhel and Apal to release the truncated DPRP-3f gene.
  • the DNA fragment carrying the DPRP-3f gene was gel band purified using the above kit and then inserted into expression vector pcDNA3.1(-)/myc- His B (Invitrogen, Carlsbad CA, USA) to make the tagged DPRP-3 expression construct ⁇ cDNA-MycHis-DPRP3.
  • RT PCR was also carried out on a number of human cell lines including but not limited to prostate cancer cells (LNCaP, PC3, DU145), the MLTC-1 line (mouse testis), and MDA-MB231 cells (breast cancer). Bands of the expected sizes for DPRP-1, DPRP-2 and DPPIV were all expressed in the various cancer cells lines, with FAP also being expressed at very low levels.
  • Northern blot analysis was performed with 2 ⁇ g poly(A) + RNA isolated from eight different tissues using DPRP probes. Specifically, a human Multiple Tissue Northern (MTN) blot (Clontech, Palo Alto, Calif.) was probed with a 1 kb N-terminal fragment that had been radioactively labeled by random priming in the presence of a 32 PdCTP (A. P. Feinberg et al, Anal. Biochem.. 132, 6 (1983)). Hybridization was performed at 68°C overnight in ExpressHybTM hybridization solution (Clontech, Palo Alto, Calif). The blots were first washed at room temperature in 2 times SSC and
  • DPRP-1 was expressed in several tissues with the most abundant signal being in testis, prostate, muscle and brain. Testis showed 3 transcripts approximately 7.5, 4.5 and 2.5 kb in length. The shorter mRNA species was very abundant in testis but negligible in the other tissues tested. DPRP-2 was ubiquitously expressed in every tissue with highest levels in liver and muscle and a predominant transcript at 5kb. DPRP-3 expression was limited to brain and pancreas. Further analysis was conducted for the three proteases in specific brain regions (cerebellum, cortex, medulla, spinal cord, occipital lobe, frontal lobe temporal lobe and putamen). DPRP-1 was expressed in all regions with low levels present in the spinal cord, while DPRP-2 was expressed in all brain regions tested.
  • Oligonucleotide primers SEQ ID NO:48 and NO:49 were used for DPRP-1 quantitative PCR, whereas oligonucleotide primers SEQ ID NO:53 and NO:54 were used for DPRP-2 quantitative PCR.
  • Human Multiple Tissue cDNA (MTCTM) Panel I and Panel II were used as normalized cDNA templates. 0.5 ng of each cDNA were used in a-25 ⁇ l PCR reaction, with each primer at a final concentration of 300 nM.
  • the PCR reaction was performed using a SYBR Green PCR Core Reagents Kit (Applied Biosystems, Foster City CA, USA) and detected with an Applied Biosystems GeneAmp 5700 sequence detection system.
  • Example 3 Production of DPRP Polyclonal Antibodies and Western Blotting The amino acid sequence deduced from the cDNA encoding DPRP-1 was analyzed using DNASTAR software (DNASTAR, Inc.) to determine regions of high immunogenicity, and a corresponding oligopeptide was synthesized and used to raise anti-DPRP- 1 antibodies.
  • oligopeptides that are about 15 to 20 residues in length, e.g. SEQ ID NO:59 for DPRP-1, SEQ ID NO:60 for DPRP-2 and SEQ ID NO:61 for DPRP-3, were synthesized using an Applied Biosystems Peptide Synthesizer Model 431 A.
  • Fmoc-chemistry was used and the 19- or 15-residue peptides were respectively coupled to keyhole limpet hemocyanin (KLH, Sigma, St. Louis, Mo.) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS).
  • KLH keyhole limpet hemocyanin
  • MBS N-maleimidobenzoyl-N-hydroxysuccinimide ester
  • Rabbits were immunized with the oligopeptide-KLH complex in complete Freund's adjuvant.
  • the resulting antisera were tested for antipeptide activity, e.g., by binding the peptide to plastic, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radioiodinated, goat anti-rabbit IgG.
  • DPRP-1 and DPRP-2 proteins were detected in brain, muscles, kidney, prostate, testis and ovary tissues.
  • DPRP-1 and DPRP-2 were synthesized as approximately lOlkDa and lOOkDa forms, respectively, which are in good agreement with the molecular masses estimated from their primary structure as shown in Table 3.
  • Table 3 also shows the number of potential N-gfycosylation sites for the DPRP proteins.
  • the presence of glycosylated and unglcosylated forms of the proteins was evaluated using tunicamycin, an inhibitor of the oligosaccharide synthesis. It is evident that the smaller forms were unglycosylated forms.
  • the correlation between mRNA (Northern analysis) and protein quantity (Western analysis) for DPRP-1 is shown in Table 4.
  • Plasmid DNA of pcDNA-DPRPl, pcDNA-MycHis-DPRPl, pcDNA-DPRP-2 or pcDNA-MycHis-DPRP2 was transfected into PEAK (EdgeBioSystems, Gaithersburg MD, USA) or COS- 1 (ATCC CRL- 1650) using LipofectAmine (Life Technologies, Gaithersburg MD, USA) method recommended by the manufacturer. Transfected cells were maintained in DMEM with 5% FBS at 37°C with 5% CO 2 for 48 hours. Cells were then collected and used for recombinant protein extraction. Cells were harvested 48 hours after transfection, homogenized and then spun at 18,000 x g for 40 min.
  • the supernata were collected as cytosolic fractions. This fraction was loaded on TALON spin column (Clontech), and His-tagged proteins were eluted with 50mM PBS, 150mM imidazole, pH 7. Recombinant proteins were then detected by western blotting with anti-myc antibody and visualized using a ProtoBlot II AP system (Promega). Recombinant affinity purified fusions of the DPRP-1 and DPRP-2 were detected by western blot, and DPRP-1 and DPRP-2 were synthesized as 112kDa and 109kDa forms as predicted.
  • Naturally occurring or recombinant DPRP proteins were substantially purified by immunoaffinity chromatography using antibodies specific for DPRP-1, DPRP-2 or DPRP-3.
  • An immunoaffinity column was constructed by covalently coupling DPRP antibodies to an activated chromatographic resin, such as CNBr-activated Sepharose (Pharmacia & Upjohn). After the coupling, the resin was blocked and washed according to the manufacturer's instructions.
  • DPRPs Media or cell extracts containing DPRP proteins were passed over the immunoaffinity column, and the column was washed under conditions that allow the preferential absorbance of DPRPs (e.g., high ionic strength buffers in the presence of detergent). The column was eluted under conditions that disrupt antibody/DPRP binding (e.g., a buffer of pH 2-3 or a high concentration of a chaotrope, such as urea or thiocyanate ion), and purified DPRP was collected.
  • a buffer of pH 2-3 or a high concentration of a chaotrope, such as urea or thiocyanate ion e.g., a buffer of pH 2-3 or a high concentration of a chaotrope, such as urea or thiocyanate ion
  • DPRP-1 and DPRP-2 were determined in a continuous fluorimetric assay. Buffer, pH and temperature dependence optimization led to the following assay conditions: Enzyme assays were performed in 50mM PBS, pH7.4 50 ⁇ l (50 ⁇ g/ml) of purified enzymes were mixed with 1 ⁇ l of different concentration of Ala-Pro-AMC (Enzyme Systems). Plates were then incubated at 37°C for 30 min, and fluorescence was detected using a Wallac 1420 Fluorimeter with ⁇ ex40355 and ⁇ em535. The K- n values of DPRP-1 and DPRP-2 were similar (208 and 161 ⁇ M respectively).
  • DPRP-1 and DPRP-2 are dipeptidyl peptidases. The data were derived by determining the fluorescence change following a 30-minute incubation of the substrates (125 ⁇ M) with enzymes as a percentage of the fluorescence measured at Ala-Pro-AMC and Gly- Pro-AMC were the only good substrates among those tested. Table 6. DPRP-1 and DPRP-2 are dipeptidyl peptidases.
  • the enzyme assay method described here is one of a number of methods that can be utilized to screen for peptide and non-peptide inhibitors of the DPRP enzymes.
  • Candidate inhibitors were prepared as 10-20 mM stock solutions in DMSO and stored at -20°C. Dilutions were made in assay buffer. Inhibition was determined by comparing the changes in fluorescence of the inhibited enzyme to the change in fluorescence of the control (vehicle) enzyme. 100-(fl units of sample/fl units of control x 100) gives percent inhibition value. The percent inhibition and the inhibitor concentration at which the enzyme was 50% inhibited (IC 50 ) was ascertained by plotting percent inhibition vs. inhibitor concentration on the log scale.
  • the DPRP-1 and DPRP-2 polypeptides to be used in the assay can be obtained by, for example, in vitro translation, recombinant expression (see Example 5) or biochemical procedures. Methods other than those described here can also be used to screen and identify compounds that inhibit DPRP-1, DPRP-2 or DPRP-3, which methods can include, for example, binding assays such as ELISAs and RIAs.
  • Example 7 Effect of DPRP Inhibitors on the Proliferation of Human Cancer Cells In Vitro
  • LNCap, PC3 and Dul45, mouse testis line MLTC-1 and MDA-MB231 breast cancer cells were plated (10 4 per well) in 96- well tissue culture plates and allowed to grow and attach for 24 hours at 37°C in a CO 2 incubator.
  • Compounds at various dilutions (final dilutions: 0.1 nM - 10 ⁇ M) were then added to the wells for various incubation periods from 24 hours to 96 hours, with fresh compound being replaced each day.
  • DPRPs are expressed in a wide variety of tissues as has been demonstrated by mRNA amplification, western blotting and immunohistochemistry.
  • DPRP-1 was most abundant in the testis by Northern blot and western blot.
  • the large number of expressed sequence tags (ESTs) from testis cDNA sources that are homologous to DPRP-1 also confirms abundant expression of DPRP-1 in testis.
  • Example 4 describes the immunohistochemical localization of DPRP-1 protein in human testis using a specific DPRP-1 antibody.
  • DPRP-1 is strongly expressed in epitheloid Leydig cells, and Leydig cells are the primary source of testicular androgens (male steroid hormones) in the mammalian male.
  • Leydig cells and macrophages are in close association with "digitation" of Leydig cell process extending onto macrophage surface. Multinucleated cells in close proximity to the Leydig cells were also stained with DPRP-1 antibody suggesting that the protease was also expressed in macrophages, and macrophages in the testis play an important role in the paracrine regulation of Leydig cells. Cytokines secreted by the testicular macrophages are mitogenic to Leydig cells and play an important role in the differentiation of mesenchymal progenitor cell into mature Leydig cells. A clearer understanding of the proteins and pathways involved in the maturation of the testis is important for the discovery of new treatments for precocious puberty. In addition,
  • Leydig cells cause tumors such as sex cord-stromal tumors via sexual steroid production (predominantly testosterone).
  • Testosterone is associated with several neoplasia and diseases such as breast carcinoma and uterine cancers, ovarian carcinoma and androgenic alopecia (hair loss).
  • Further examination of the localization of DPRP proteins in other glands in the body (e.g. adrenal glands) that produce testosterone and other androgenic hormones are currently under investigation.
  • the possible association of DPRP-1 with steroid and polypeptide hormone biosynthetic pathways functions is being investigated, and Example 7 is relevant to understanding the role of DPRP proteins in prostate, testis and breast in vitro cell models.
  • Immunohistochemical analysis also localized DPRP-1 to endometrial glands in the uterus (see Example 4), pancreatic acini, glomeruli of the kidney, plasma cells in the bladder, a subset of B-cells in the tonsils, columnar epithelial cells of the prostate and poorly differentiated prostate squamous metaplasia, Gleason grade 4 prostatic carcinoma, and hyperplastic glands in benign prostatic hyperplasia.
  • Positive staining in breast carcinoma, as well as in seminoma and prostate squamous metaplasia suggests a general association of DPRP-1 with hormone-sensitive tissues, particularly in cells that become poorly differentiated.
  • DPRP-1 inflammatory breast carcinoma has an abundance of infiltrating lymphocytes and an overall bad prognosis.
  • DPRP-1 and other DPRP proteins appear in medullary carcinomas that typically have a constant infiltrating lymphoplasmacytic component at the periphery of the tumor, which is thought to represent a reaction of the host tissues to the neoplasm.
  • Most of the lymphocytes are T Cells, and most of the plasma cells are of the IgG-producing type.
  • DPRP-1 in specialized epithelial cells of a number of tissues suggests that DPRP-1 and other DPRP proteins may be involved in growth and differentiation thereof.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Neurology (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Neurosurgery (AREA)
  • Diabetes (AREA)
  • Oncology (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Communicable Diseases (AREA)
  • Virology (AREA)
  • Obesity (AREA)
  • Endocrinology (AREA)
  • Vascular Medicine (AREA)
  • Pain & Pain Management (AREA)

Abstract

L'invention concerne de nouveaux polypeptides ou protéines possédant une forte homologie de séquence avec la dipeptidylpeptidase IV (DPPIV), et par conséquent des acides nucléiques codant pour ces protéines, des cellules qui ont été modifiées par lesdits acides nucléiques pour exprimer lesdites protéines, des anticorps correspondant auxdites protéines, des méthodes de sélection destinées à découvrir de nouveaux agents thérapeutiques inhibiteurs de l'activation desdites protéines ou de protéines s'y apparentant, des agents thérapeutiques découverts par lesdites méthodes de sélection, ainsi que de nouveaux traitements thérapeutiques.
EP01981501A 2000-10-12 2001-10-12 Nouveaux genes de serine protease apparentes a la dppiv Ceased EP1346033A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US24011700P 2000-10-12 2000-10-12
US240117P 2000-10-12
PCT/US2001/031874 WO2002031134A2 (fr) 2000-10-12 2001-10-12 Nouveaux genes de serine protease apparentes a la dppiv

Publications (1)

Publication Number Publication Date
EP1346033A2 true EP1346033A2 (fr) 2003-09-24

Family

ID=22905181

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01981501A Ceased EP1346033A2 (fr) 2000-10-12 2001-10-12 Nouveaux genes de serine protease apparentes a la dppiv

Country Status (18)

Country Link
US (2) US6844180B2 (fr)
EP (1) EP1346033A2 (fr)
JP (1) JP2004528812A (fr)
KR (1) KR100875221B1 (fr)
CN (2) CN101270362A (fr)
AU (2) AU2002213138B2 (fr)
CA (1) CA2425001A1 (fr)
CZ (1) CZ20031301A3 (fr)
HK (1) HK1078104A1 (fr)
HU (1) HUP0301356A3 (fr)
IL (2) IL155245A0 (fr)
MX (1) MXPA03003191A (fr)
NO (1) NO329842B1 (fr)
NZ (1) NZ525443A (fr)
PL (1) PL366005A1 (fr)
RU (1) RU2305133C2 (fr)
WO (1) WO2002031134A2 (fr)
ZA (1) ZA200303306B (fr)

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6979697B1 (en) * 1998-08-21 2005-12-27 Point Therapeutics, Inc. Regulation of substrate activity
JP4748908B2 (ja) * 1999-09-10 2011-08-17 ザ・ユニバーシティ・オブ・シドニー ジペプチジルペプチダーゼ
US20040047853A1 (en) * 2000-06-09 2004-03-11 Dahl Soren W Purified proenzyme of dipeptidyl peptidase i (pro-dppi)
AUPR107800A0 (en) * 2000-10-27 2000-11-23 University Of Sydney, The Peptide and nucleic acid molecule ii
GB0101760D0 (en) * 2001-01-23 2001-03-07 Glaxo Group Ltd Novel protein
US20050123910A1 (en) * 2001-04-24 2005-06-09 Cookson William Osmond C.M. Enzyme and snp marker for disease
AU2002308637B2 (en) 2001-05-11 2007-05-17 Board Of Regents, The University Of Texas System Anti-cd26 monoclonal antibodies as therapy for diseases associated with cells expressing cd26
DE10150203A1 (de) 2001-10-12 2003-04-17 Probiodrug Ag Peptidylketone als Inhibitoren der DPIV
US6709651B2 (en) * 2001-07-03 2004-03-23 B.M.R.A. Corporation B.V. Treatment of substance P-related disorders
WO2003057916A2 (fr) * 2002-01-09 2003-07-17 Riken Profils du cancer
DK1528931T3 (da) 2002-08-09 2008-09-08 Prosidion Ltd Dipeptidylpeptidase-IV-inhibitorer til reduktion af hastigheden af kronisk vægtforögelse
CN1894234A (zh) 2003-03-25 2007-01-10 武田药品工业株式会社 二肽基肽酶抑制剂
JP2007511467A (ja) 2003-05-14 2007-05-10 タケダ サン ディエゴ インコーポレイテッド ジペプチジルペプチダーゼインヒビター
US7723344B2 (en) 2003-08-13 2010-05-25 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
US7678909B1 (en) 2003-08-13 2010-03-16 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
US7169926B1 (en) 2003-08-13 2007-01-30 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
EP1699777B1 (fr) 2003-09-08 2012-12-12 Takeda Pharmaceutical Company Limited Inhibiteurs de la dipeptidylpeptidase
US7732446B1 (en) 2004-03-11 2010-06-08 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
CN102134230B (zh) 2004-03-15 2019-06-28 武田药品工业株式会社 二肽基肽酶抑制剂
EP1745293A1 (fr) * 2004-04-28 2007-01-24 Bayer HealthCare AG Agents utilises dans le diagnostic et le traitement des maladies associees a la dipeptidyl-peptidase 9 (dpp9)
WO2005106021A1 (fr) * 2004-04-28 2005-11-10 Bayer Healthcare Ag Diagnostics et therapeutique de maladies associees a la dipeptidyle peptidase 8 (dpp8)
US7687638B2 (en) 2004-06-04 2010-03-30 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
WO2006019965A2 (fr) 2004-07-16 2006-02-23 Takeda San Diego, Inc. Inhibiteurs de la dipeptidyl peptidase
US20060063719A1 (en) * 2004-09-21 2006-03-23 Point Therapeutics, Inc. Methods for treating diabetes
EP1828192B1 (fr) * 2004-12-21 2014-12-03 Takeda Pharmaceutical Company Limited Inhibiteurs de dipeptidyle peptidase
SI1942898T2 (sl) 2005-09-14 2014-08-29 Takeda Pharmaceutical Company Limited Dipeptidil-peptidazni inhibitorji za zdravljenje diabetesa
PE20070622A1 (es) * 2005-09-14 2007-08-22 Takeda Pharmaceutical Administracion de inhibidores de dipeptidil peptidasa
CN102675221A (zh) 2005-09-16 2012-09-19 武田药品工业株式会社 用于制备嘧啶二酮衍生物的方法中的中间体
WO2007047205A2 (fr) * 2005-10-11 2007-04-26 Amano Enzyme Usa Co., Ltd. Inhibiteurs enzymatiques du pai-1
WO2007054577A1 (fr) 2005-11-14 2007-05-18 Probiodrug Ag Derives de pyrrolidine a fusion de cyclopropyle, en tant qu'inhibiteurs de dipeptidyle peptidase iv (dp iv)
WO2007112347A1 (fr) 2006-03-28 2007-10-04 Takeda Pharmaceutical Company Limited Inhibiteurs de la dipeptidyl peptidase
BRPI0709984A2 (pt) 2006-04-12 2011-08-02 Probiodrug Ag inibidores de enzima
US8324383B2 (en) 2006-09-13 2012-12-04 Takeda Pharmaceutical Company Limited Methods of making polymorphs of benzoate salt of 2-[[6-[(3R)-3-amino-1-piperidinyl]-3,4-dihydro-3-methyl-2,4-dioxo-1(2H)-pyrimidinyl]methyl]-benzonitrile
TW200838536A (en) 2006-11-29 2008-10-01 Takeda Pharmaceutical Polymorphs of succinate salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethy]-4-fluor-benzonitrile and methods of use therefor
CN101259119B (zh) * 2007-03-08 2010-05-19 上海市计划生育科学研究所 一种丝氨酸蛋白酶抑制剂及其衍生物在生育调节中的应用
US8093236B2 (en) 2007-03-13 2012-01-10 Takeda Pharmaceuticals Company Limited Weekly administration of dipeptidyl peptidase inhibitors
WO2014068023A1 (fr) * 2012-11-02 2014-05-08 Georg-August-Universität Göttingen Stiftung Öffentlichen Rechts, Universitätsmedizin Inhibiteurs peptidiques de dpp8 et dpp9
US11427814B2 (en) 2019-03-26 2022-08-30 Encodia, Inc. Modified cleavases, uses thereof and related kits
WO2020198264A1 (fr) * 2019-03-26 2020-10-01 Encodia, Inc. Clivages modifiés, utilisations et kits correspondants

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6291662B1 (en) 1984-12-05 2001-09-18 Amgen Inc. Recombinant methods for production of serine protease inhibitors and DNA sequences
IL111785A0 (en) 1993-12-03 1995-01-24 Ferring Bv Dp-iv inhibitors and pharmaceutical compositions containing them
CA2360464A1 (fr) 1999-01-11 2000-07-20 Incyte Pharmaceuticals, Inc. Peptidases humaines
JP2004507202A (ja) 1999-03-31 2004-03-11 キュラジェン コーポレイション ポリペプチドをコードするオープンリーディングフレームを含む核酸;「orfx」
JP4748908B2 (ja) * 1999-09-10 2011-08-17 ザ・ユニバーシティ・オブ・シドニー ジペプチジルペプチダーゼ
CN1307128A (zh) * 2000-01-26 2001-08-08 上海博道基因技术有限公司 一种新的多肽——人二肽氨肽酶28和编码这种多肽的多核苷酸
CA2411971A1 (fr) * 2000-06-16 2001-12-27 Incyte Genomics, Inc. Proteases

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0231134A2 *

Also Published As

Publication number Publication date
WO2002031134A2 (fr) 2002-04-18
ZA200303306B (en) 2004-08-12
RU2305133C2 (ru) 2007-08-27
IL155245A0 (en) 2003-11-23
IL155245A (en) 2010-04-15
HK1078104A1 (en) 2006-03-03
US6844180B2 (en) 2005-01-18
PL366005A1 (en) 2005-01-24
CA2425001A1 (fr) 2002-04-18
US20050059081A1 (en) 2005-03-17
NO20031702L (no) 2003-05-15
CZ20031301A3 (cs) 2003-10-15
US7157241B2 (en) 2007-01-02
KR100875221B1 (ko) 2008-12-19
JP2004528812A (ja) 2004-09-24
AU1313802A (en) 2002-04-22
AU2002213138B2 (en) 2006-07-20
NZ525443A (en) 2006-04-28
NO20031702D0 (no) 2003-04-11
MXPA03003191A (es) 2004-12-03
NO329842B1 (no) 2011-01-10
CN101270362A (zh) 2008-09-24
CN100354417C (zh) 2007-12-12
KR20030038815A (ko) 2003-05-16
CN1636061A (zh) 2005-07-06
US20020115843A1 (en) 2002-08-22
HUP0301356A2 (hu) 2003-10-28
HUP0301356A3 (en) 2010-03-29
WO2002031134A3 (fr) 2003-07-17

Similar Documents

Publication Publication Date Title
US6844180B2 (en) Serine protease genes related to DPPIV
AU2002213138A1 (en) Novel serine protease genes related to DPPIV
CA2443236A1 (fr) Cathepsine derivee d'ostoclaste humain
US7115260B2 (en) Interleukin-1β converting enzyme like apoptotic protease-6
JP2002501739A (ja) ヒトシスタチンf
CA2464343A1 (fr) Proteines isolees secretees par l'humain, molecules d'acides nucleiques codant pour ces proteines secretees par l'humain et leur utilisation
JP2002191379A (ja) Asp1
JPH1175873A (ja) 新規化合物
US20020090373A1 (en) ADAMTS polypeptides, nucleic acids encoding them, and uses thereof
KR20070116932A (ko) 뉴로트립신의 억제제
EP0861898A1 (fr) Polynucleotide et séquence de polypeptide la Cathepsine-K du rat
US6358702B1 (en) Polynucleotides encoding human Hox C10
AU2006228068B2 (en) Novel serine protease genes related to DPPIV
JPH1175874A (ja) 新規化合物
NZ542643A (en) Serine protease genes related to Dipeptidyl peptidase IV
EP0846763A2 (fr) R5, une protéine se liant à la protéine phosphatase-1
WO2002066050A1 (fr) Inhibiteurs de caspase 3
EP1069188A1 (fr) Métallopeptidases membranaires de type neprolysin
JP2001186889A (ja) 良性前立腺肥大と関連するポリヌクレオチドおよびポリペプチド
US6890721B1 (en) Interleukin-1β converting enzyme like apoptotic protease-6
JP2000083683A (ja) Frzbファミリ―のメンバ―、frazzled
JP2002191376A (ja) ヒト・レクイエム
JP2002085059A (ja) 新規なtsp1ドメイン含有ポリペプチド
JPH11505110A (ja) ヒトニューロペプチドレセプター

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20030506

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

17Q First examination report despatched

Effective date: 20060725

17Q First examination report despatched

Effective date: 20060725

APBK Appeal reference recorded

Free format text: ORIGINAL CODE: EPIDOSNREFNE

APBN Date of receipt of notice of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA2E

APBR Date of receipt of statement of grounds of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA3E

APAF Appeal reference modified

Free format text: ORIGINAL CODE: EPIDOSCREFNE

APAF Appeal reference modified

Free format text: ORIGINAL CODE: EPIDOSCREFNE

REG Reference to a national code

Ref country code: DE

Ref legal event code: R003

APBT Appeal procedure closed

Free format text: ORIGINAL CODE: EPIDOSNNOA9E

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20141024